Comparative oncology as a tool in drug development requires a deeper examination of the value of the approach and examples of where this approach can satisfy unmet needs. This review seeks to demonstrate types of drug development questions that are best answered by the comparative oncology approach. We believe common perceived risks of the comparative approach relate to uncertainty of how regulatory bodies will prioritize or react to data generated from these unique studies conducted in diseased animals, and how these new data will affect ongoing human clinical trials. We contend that it is reasonable to consider these data as potentially informative and valuable to cancer drug development, but as supplementary to conventional preclinical studies and human clinical trials particularly as they relate to the identification of drug-associated adverse events. Clin Cancer Res; 22(9); 2133–8. ©2015 AACR.

The study of naturally occurring cancer in companion animals, known as comparative oncology, forms the basis of a translational drug development strategy that primarily includes tumor-bearing pet dogs in clinical trials of novel cancer therapies destined for use in human cancer patients (1–5). The recognition of spontaneous cancer development in companion animals, and potential for inclusion of such animals in drug development studies, is based upon observations of canine malignancies that share morphologic, histologic, and biologic characteristics with human cancers. Dogs' physical size, amenability to serial biologic sample collections, compressed survival compared with humans, comparable tumor biology, intact immunity, and relevant responses to cytotoxic therapies provide clear support to their inclusion as a complementary animal model (4, 5).

Currently, the field of comparative oncology is focused on tumor-bearing dogs as they comprise the majority of those presented to veterinarians for cancer diagnosis and management, which is in turn facilitated by scientific knowledge of malignancies they develop, the collective veterinary clinical experience with anticancer therapies such as chemotherapy and radiation, and availability of basic annotation of the canine genome and immune system. A major milestone was establishment of the NCI's Comparative Oncology Program (NCI-COP) at the NIH in 2004. A component of this program is the Comparative Oncology Trials Consortium (NCI-COTC; https://ccrod.cancer.gov/confluence/display/CCRCOPWeb/Home), an infrastructure uniting study sponsors, such as pharmaceutical and biotechnology companies, with 21 academic veterinary centers within North America to support multicenter clinical trials of investigational therapeutics, wherein centralized trial support and data management is provided by the NCI (6, 7). This mechanism provides access to a clinical trial infrastructure that delivers trial results in a facile manner, considerate of timelines generally required in drug development strategies. Furthermore, a body of published work now exists to demonstrate the feasibility and applicability of the dog cancer model in drug development to ensure data that are both scientifically sound and robust, thus supporting inclusion into FDA applications. Although not formal FDA guidance, direction for clinical trial conduct and data reporting exists for drugs evaluated in comparative oncology studies in the pre- and post-Investigational New Drug (IND) settings, and has been used effectively by groups actively involved in these efforts (8).

Today's challenge is how to best capture and convey the value of these studies, given the timeline for drug development and the diversity of data that collectively informs decisions in the development path. Various attempts at defining value have been made, including a financial model that proposes savings of billions of research and development dollars, achieved primarily through the effective design of better phase II human studies (9). We propose that the value of the comparative approach lies in the answers to critical drug development questions that are not answered in human trials or conventional preclinical models. Herein, we present a summary of the types of questions that are best asked and answered by comparative oncology studies (Table 1), along with a discussion of selected studies that generated answers to such questions, thus are demonstrative of the value of the comparative oncology approach.

Table 1.

Examples of drug development questions that can be answered within the comparative oncology approach

QuestionOutcomeSelected supporting references
Safety and efficacy 
What is the clinical response to an investigational agent, and can this response be characterized using standardized, quantitative metrics, including imaging techniques, that are translatable to human clinical studies? Common criteria employed to communicate tumor responses between veterinary and human patients 35–40 
What is the success of an investigational agent in the context of treatment-naïve disease? Ability to observe responses in patients without preexisting drug resistance as seen in human phase I 41–45 
What is the acute and chronic toxicity profile of an investigational agent, both as a single agent and in combination with conventional chemotherapy? Can this be described with standardized metrics? Common criteria employed to communicate tumor responses between veterinary and human patients; provide insight into what to expect/monitor for within human patients. 10, 11, 14, 15, 41, 45 
Which histologies appear to be most likely to respond to a specific investigational drug or drug class? Insight into comparable tumor types for study within human patients 10, 11 
Pharmacokinetics/pharmacodynamics 
What are the relationships between therapeutic index, demonstration of pharmacodynamics endpoints, and tolerable drug exposure? Provides supporting data to select a biologically effective dose in conjunction with or instead of an MTD 10, 11, 23, 24, 45, 46 
Can pharmacokinetic/pharmacodynamic data obtained in dogs be used to define the optimal dose and schedule of a new drug? Optimization of dose/schedule prior to prescription of RP2 dose in humans; identification of MTD in dogs 10, 11, 42, 45 
Can differential pharmacokinetic/pharmacodynamic relationships within blood versus tumor be characterized to identify which biologic sample is most indicative of pharmacodynamic effect? Ability to tailor biologic sample collection and correlative assay development for human trials 12 
Given a comparable therapeutic index of a given drug between humans and dogs, do therapeutically relevant levels of drug accumulate in tumors at a given (tolerable) dose, schedule, and pharmacokinetic profile? Provides further proof-of-principle or lack thereof for systemically administered drugs across a range of doses NCI Comparative Oncology Trials Consortium (COTC): A clinical trial of iniparib in tumor-bearing dogs (in press) 
Drug target investigations 
Can comparative cancer studies in tumor-bearing dogs identify potential new targets that are druggable in both dogs and humans, and/or identify new molecular signatures that correlate with prognosis? New candidate genes identified from dog data to support investigations in human patients; shared molecular derangements identified to add comparative relevance to the canine model 15 
Can biospecimen repositories be populated with sufficient canine samples to allow matched tissues from primary and metastatic sites within the same patient to allow elucidation of drug targets within the metastatic pathway? Provides an unparalleled high-quality resource for canine comparative cancer biology investigations 26, 27 
Imaging agent validation 
Can imaging agent performance and validation (target:background ratios, off-target binding, normal biodistribution, lesions distribution kinetics) be assessed in canine cancer patients? Validated imaging signal against clinical findings and tissue histology; allowed exploration of imaging agent dose and subsequent performance to be validated both in vivo and with ex vivo tissue imaging taken during surgical procedures 17, 39, 47, 48, 49 
Can validation of novel imaging agents or feasibility of new imaging protocols be assessed in canine cancer patients? Allowed exploration of novel combinations of different imaging agents, particularly those with variable radiopharmaceutical composition 17, 50–54 
Nontraditional study design   
How can a drug's performance in the minimal residual disease (MRD) setting be assessed? Can clinical endpoints that do not involve regression of clinically measurable lesions, rather delay of onset of metastasis, be evaluated? Provided an opportunity to assess impact on disease-free interval after removal of a primary tumor with a known high risk of metastatic progression 55 
Can the dog serve as a model for personalized medicine studies to validate processes? Validated a workflow for prospective genomic profiling of individual canine tumors, enabling future basket trial designs 56 
Biomarkers 
How can biomarkers of drug exposure in both normal (plasma, PBMCs) and tumor tissues be identified and validated? Demonstrates the strengths of the canine cancer model for collection of biologic samples within various compartments (blood, tumor tissue, normal tissue) 7, 23, 24, 26 
Can new biomarkers for prognosis be identified from dogs receiving a novel drug? Showed baseline expression of key tumoral factors, such as necrosis, inflammation was prognostic for clinical outcome 47, 57 
Can immune response data be collected from dogs receiving tumor-specific immunotherapy that is correlative and/or predictive of tumor response? Demonstrates the availability and utility of immune cell assays as valid pharmacodynamic readout for immune-based therapies 15, 30 
Prioritization of candidate drugs 
Can comparative oncology studies play a role in lead candidate drug selection for advancement in the human clinic? Provided robust correlative data to enhance the preclinical data package to discern and prioritize candidates for human use NCI Comparative Oncology Trials Consortium (COTC): Indeno-isoquinoline candidates in canine lymphoma (in progress) 
QuestionOutcomeSelected supporting references
Safety and efficacy 
What is the clinical response to an investigational agent, and can this response be characterized using standardized, quantitative metrics, including imaging techniques, that are translatable to human clinical studies? Common criteria employed to communicate tumor responses between veterinary and human patients 35–40 
What is the success of an investigational agent in the context of treatment-naïve disease? Ability to observe responses in patients without preexisting drug resistance as seen in human phase I 41–45 
What is the acute and chronic toxicity profile of an investigational agent, both as a single agent and in combination with conventional chemotherapy? Can this be described with standardized metrics? Common criteria employed to communicate tumor responses between veterinary and human patients; provide insight into what to expect/monitor for within human patients. 10, 11, 14, 15, 41, 45 
Which histologies appear to be most likely to respond to a specific investigational drug or drug class? Insight into comparable tumor types for study within human patients 10, 11 
Pharmacokinetics/pharmacodynamics 
What are the relationships between therapeutic index, demonstration of pharmacodynamics endpoints, and tolerable drug exposure? Provides supporting data to select a biologically effective dose in conjunction with or instead of an MTD 10, 11, 23, 24, 45, 46 
Can pharmacokinetic/pharmacodynamic data obtained in dogs be used to define the optimal dose and schedule of a new drug? Optimization of dose/schedule prior to prescription of RP2 dose in humans; identification of MTD in dogs 10, 11, 42, 45 
Can differential pharmacokinetic/pharmacodynamic relationships within blood versus tumor be characterized to identify which biologic sample is most indicative of pharmacodynamic effect? Ability to tailor biologic sample collection and correlative assay development for human trials 12 
Given a comparable therapeutic index of a given drug between humans and dogs, do therapeutically relevant levels of drug accumulate in tumors at a given (tolerable) dose, schedule, and pharmacokinetic profile? Provides further proof-of-principle or lack thereof for systemically administered drugs across a range of doses NCI Comparative Oncology Trials Consortium (COTC): A clinical trial of iniparib in tumor-bearing dogs (in press) 
Drug target investigations 
Can comparative cancer studies in tumor-bearing dogs identify potential new targets that are druggable in both dogs and humans, and/or identify new molecular signatures that correlate with prognosis? New candidate genes identified from dog data to support investigations in human patients; shared molecular derangements identified to add comparative relevance to the canine model 15 
Can biospecimen repositories be populated with sufficient canine samples to allow matched tissues from primary and metastatic sites within the same patient to allow elucidation of drug targets within the metastatic pathway? Provides an unparalleled high-quality resource for canine comparative cancer biology investigations 26, 27 
Imaging agent validation 
Can imaging agent performance and validation (target:background ratios, off-target binding, normal biodistribution, lesions distribution kinetics) be assessed in canine cancer patients? Validated imaging signal against clinical findings and tissue histology; allowed exploration of imaging agent dose and subsequent performance to be validated both in vivo and with ex vivo tissue imaging taken during surgical procedures 17, 39, 47, 48, 49 
Can validation of novel imaging agents or feasibility of new imaging protocols be assessed in canine cancer patients? Allowed exploration of novel combinations of different imaging agents, particularly those with variable radiopharmaceutical composition 17, 50–54 
Nontraditional study design   
How can a drug's performance in the minimal residual disease (MRD) setting be assessed? Can clinical endpoints that do not involve regression of clinically measurable lesions, rather delay of onset of metastasis, be evaluated? Provided an opportunity to assess impact on disease-free interval after removal of a primary tumor with a known high risk of metastatic progression 55 
Can the dog serve as a model for personalized medicine studies to validate processes? Validated a workflow for prospective genomic profiling of individual canine tumors, enabling future basket trial designs 56 
Biomarkers 
How can biomarkers of drug exposure in both normal (plasma, PBMCs) and tumor tissues be identified and validated? Demonstrates the strengths of the canine cancer model for collection of biologic samples within various compartments (blood, tumor tissue, normal tissue) 7, 23, 24, 26 
Can new biomarkers for prognosis be identified from dogs receiving a novel drug? Showed baseline expression of key tumoral factors, such as necrosis, inflammation was prognostic for clinical outcome 47, 57 
Can immune response data be collected from dogs receiving tumor-specific immunotherapy that is correlative and/or predictive of tumor response? Demonstrates the availability and utility of immune cell assays as valid pharmacodynamic readout for immune-based therapies 15, 30 
Prioritization of candidate drugs 
Can comparative oncology studies play a role in lead candidate drug selection for advancement in the human clinic? Provided robust correlative data to enhance the preclinical data package to discern and prioritize candidates for human use NCI Comparative Oncology Trials Consortium (COTC): Indeno-isoquinoline candidates in canine lymphoma (in progress) 

Small molecules and the relationship of pharmacokinetics, pharmacodynamics, and clinical assessment of tolerability and efficacy

A highly soluble prodrug of ganetespib, STA-1474, was studied in dogs with cancer to establish clinical toxicity, to identify surrogate biomarkers of response and pharmacokinetics between two proposed dosing schedules, and to provide evidence of biologic activity. This study met all defined objectives, and assisted in devising a dosing strategy to provide prolonged drug exposure to support efficient inhibition of drug target via modulation of a surrogate biomarker in blood [HSP70 upregulation in peripheral blood mononuclear cells (PBMC)] and tumor levels of c-kit (an HSP90 client protein). Collectively, this data informed the design of human clinical trials of ganetespib and demonstrates the strengths of a naturally occurring canine model by highlighting the ease of serial biopsy procurement, rapid assessment of differential dose and schedule, and correlative assessment of multiple clinical parameters (10). In another similar example, an orally bioavailable XPO1 inhibitor verdinexor, a companion agent to a lead human compound KPT-330 (Selinexor, Karyopharm Therapeutics), was studied in tumor-bearing dogs. Based upon profound clinical benefit observed in dogs with non-Hodgkin lymphoma (NHL) and the marked similarities between canine and human NHL, the data generated within this study provided critical new information in support of related compounds in humans with hematologic malignancies (11). Selinexor is currently being evaluated in phase I and II clinical trials for a variety of human cancers.

Biomarker validation and optimization of pharmacodynamics assays within the context of drug exposure in tumor-bearing dogs

The irreversible inhibitor of Bruton tyrosine kinase (Btk), ibrutinib, was studied in dogs with B-cell lymphoma to establish tolerability, preliminary efficacy data, and to validate a pharmacodynamic assay within PBMCs and tumor tissue. Validation of the fluorescently labeled derivative of ibrutinib to monitor occupancy of Btk by the drug has led to adoption of this approach as pharmacodynamic readout in subsequent human trials, while also supporting the use of ibrutinib in humans with B-cell malignancies (12, 13).

Another valuable example is study of hydroxychloroquine, an autophagy inhibitor, given to dogs with NHL. The pharmacodynamic response evaluated in both PBMCs and tumor tissue, obtained via serial peripheral lymph node biopsies, demonstrated that reliance on surrogate PBMC for demonstration of sufficient drug levels for effective autophagy inhibition within the tumor tissue cannot always be inferred, thus underscoring the strength of the canine cancer model (14).

Immune-modulating agents for cancer therapy

A comparative oncology study of an immunocytokine, NHS-IL12, administered subcutaneously to dogs with malignant melanoma was conducted to identify tolerability and immunologic activity of this agent across a range of doses (15). Pharmacokinetic and pharmacodynamic endpoints, in the form of serum IFNγ, IL10 levels, and intratumoral CD8+ lymphocytes, were assessed alongside clinical measures of response and toxicity. This study provided data that directly informed the design of the ongoing phase I human trial of this agent (NCT01417546). The study demonstrated both initial safety and efficacy signals in a relevant species bearing a naturally arising tumor. This data was crucial to the rigorous scientific review of the clinical trial at the NCI Center for Cancer Research (NCI/CCR) and facilitated the CCR holding the IND for this agent at a time when the study sponsor had deprioritized this compound. This currently is now a high priority agent for planned combination studies in man.(James Gulley, MD; personal communication).

Comparative cancer genomics

The use of dogs in cross-species genomics studies provides a unique opportunity to identify regions of potentially shared and clinically relevant genomic changes. In one such example, candidate genes IL8 and SLC1A3 were identified in canine osteosarcoma as overexpressed; these same genes had variable expression in human osteosarcoma but nevertheless were associated with a poor clinical outcome (16). Additional studies adopting this line of investigation could help identify yet-characterized candidate genes and/or pathways for future application to human cancer genomic studies.

Preclinical assessment of cancer imaging agents in tumor-bearing dogs

Dogs with measurable malignancies that are considered surgical candidates represent a unique opportunity to assess intraoperative imaging agents to provide, in real time, an assessment of surgical margins to inform on the extent of resection and thus optimize outcome. BLZ-100, a near-infrared imaging agent currently in phase I human studies, is a peptide–fluorophore conjugate that was evaluated in a comparative oncology study of dogs with a variety of cancer histologies. (17; Blaze Bioscience, Inc.) Canine tissues were imaged both in vivo and ex vivo to identify an efficacious dose of BLZ-100. These data provided a foundation and rationale to assess performance of this agent in human patients with soft-tissue sarcomas.

Challenges and perceived risks to comparative oncology studies

Perspectives from those within the pharmaceutical industry against using a comparative oncology approach generally include the relatively higher cost of comparative oncology trials compared with other conventional animal models, the greater amount of drug needed for dosing of dogs, and the time needed to complete such trials from inception to analysis of data. Responses to these points must include consideration of the uniqueness of the data generated within heterogeneous, spontaneous cancer that develops within an immunocompetent host, which is not generated with the intent of describing toxicity as a primary endpoint. The amount of drug and time to execute the studies should be considered in context of when the data is desired within an individual drug's development. Good Manufacturing Practices (GMP)-level material is not generally needed for comparative oncology clinical studies, although basic purity and release criteria have been previously described (7, 18). Capitalization on a multicenter clinical trial consortium such as the NCI-COTC can assist in hastening the conduct of a trial, but interim analyses will introduce natural and important pauses within the study timeline. Nevertheless, the timeline for comparative oncology studies are much shorter than typical phase I/II human trials and are conducted at a much lower cost, while providing data that can directly inform the design of human trials, representing a valuable return on investment.

Similarly important to consider are questions that cannot be effectively asked within a dog model. It is important to note that prior to initiation of comparative oncology drug trials in dogs, consideration of existing normal dog toxicology data, generated in most cases by the study sponsor during toxicologic assessment, is important to proper and ethical design of the trial. In cases where the dog is a known sensitive species for severe toxicity, and no reasonable margin of safety can be applied to demonstrate therapeutic efficacy, the tumor-bearing dog would not be appropriate for exploration of potential pharmacokinetic/pharmacodynamic relationships and how they correlate to clinical efficacy and tolerability.

The field of cancer genetics and genomics is rapidly evolving, with particular emphasis on specific knowledge of druggable pathways that are critically linked to malignant behavior, supporting the ongoing development of targeted therapies. Indeed, a deeper knowledge of the naturally occurring canine cancer genomic landscape is crucial to defining the pertinent questions that can be asked within canine cancer patients, and how relatable canine cancers are to human cancers on the genomic level. The comparative chromosome alignment technique and the differential organization of the dog genome may narrow key regions of the genome associated with cancers. Recent work in this area demonstrates that recurrent aberrations correlate with cancer subtype, and that corresponding cytogenetic lesions may exist in human patients (16, 19–22). Several examples of where the value of a cross-species approach to cancer genomics has been demonstrated exist in the literature. For example, recent work in canine melanoma demonstrates that although canine tumors possess rare mutations in BRAF and NRAS, they exhibit similar differential gene expression changes to human melanoma within downstream MAPK and PI3K/AKT pathways (25). Thus, although the driving mutations between human and canine melanoma may differ, similar activation and sensitivity to inhibition of such shared signaling pathways underscores the translational value of studying comparative melanoma biology. This aspect of comparative oncology will continue to develop and could support initiation of so-called “basket” trials wherein response of tumors with shared, credentialed biology to a specific targeted therapy are assessed, agnostic of histologic diagnosis. To facilitate future studies in this area, high-quality biologic samples from dogs with various malignancies are available via the Pfizer-Canine Comparative Oncology and Genomics Consortium (CCOGC) biospecimen repository (www.ccogc.net refs. 26, 27). This resource is uniquely suited to provide the necessary molecular background that is currently missing from the comparative oncology armamentarium. CCOGC samples are treatment-naïve, clinically annotated, and include both tumor and matched normal tissues, including peripheral blood, urine, plasma, and serum. Seven histologies were selected based upon their translational relevance at the time the resource was populated (2007–2011), totaling 1,800 individual patients and approximately 60,000 samples.

A rapidly growing field in cancer drug development is the conception and creation of biologic agents that affect an antitumor response via immune response manipulation and/or reprogramming within individual patients. For such an approach, the type of the agent may critically influence the applicability of the dog model for ongoing development. Incomplete knowledge of shared tumor antigens between humans and dogs may limit the questions that can be asked of such agents that are destined for human use. Furthermore, even with successful targeting of a specific tumor-associated antigen shared between humans and dogs, immunocompetent canine cancer patients will effectively clear any foreign (human or murine) antibodies, thus potentially limiting use of a dog model for evaluation of mAbs intended for repeated therapeutic dosing schemes unless an equivalent canin-ized or canine chimeric product is manufactured alongside the parent compound. Tumor vaccines, particularly those which rely on a shared tumor antigen(s) may be viable candidates for validation studies in a dog model (28, 29). Success in autologous tumor cell lysate vaccination strategies in canine meningioma, melanoma, and lymphoma and others have provided insight for comparative human trials (30–32). Similarly, oncolytic viruses, which capitalize on malignant cells' defects in viral response gene pathways, may be effectively translated between dogs and humans (33, 34).

A path forward: a reasonable regulatory response has been provided

During discussion at an Institute of Medicine (IOM) meeting on June 9, 2015, which included individuals from FDA, NIH, and various academic and industry stakeholders in cancer drug development, Dr. John Leighton of the FDA's Center for Drug Evaluation and Research (CDER) provided public insight into regulatory review of comparative oncology data, of which a summary is available within the IOM proceedings (http://www.nap.edu/catalog/21830/the-role-of-clinical-studies-for-pets-with-naturally-occurring-tumors-in-translational-cancer-research). Although these comments are not considered formal regulatory guidance, their importance is underscored here:

  1. The FDA is aware of the role of pet dogs may play as research subjects in human drug development settings. Such data collected in dogs is expected to be filed under the IND as it becomes available. A New Animal Drug (NAD) application is not needed for clinical studies evaluating drugs in this specific research setting.

  2. Data collected in the context of a comparative oncology clinical trial setting would be viewed in context. The FDA is aware that companion dogs are housed and treated in the home environment and may have comorbidities reflective of their age and naturally occurring malignancies.

  3. Data collected from tumor-bearing pet dogs would never “trump” human data, particularly with respect to drug tolerability and clinical response.

  4. Over the past 15 years, safety signals have never been identified in tumor-bearing pet dogs that have resulted in a clinical hold for an existing human IND study.

The questions asked and answered within comparative oncology studies are informative, unique, and not easily provided by conventional preclinical models or by most human trials. These data do not replace controlled toxicokinetic studies in purpose-bred dogs and other laboratory animal species. Regulatory review of these data would include consideration of context and the recognized complexities of working within a naturally occurring disease model system. It is imperative that investigators actively engaged in comparative oncology studies both report and characterize unexpected adverse events they observe so as to understand and attribute these events fully.

C. Khanna is an employee of Animal Clinical Investigation and Ethos Veterinary Health, and has ownership interests (including patents) at The Oncology Service. No potential conflicts of interest were disclosed by the other authors.

Conception and design: A.K. LeBlanc, C.N. Mazcko, C. Khanna

Development of methodology: A.K. LeBlanc, C. Khanna

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): A.K. LeBlanc

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A.K. LeBlanc

Writing, review, and/or revision of the manuscript: A.K. LeBlanc, C.N. Mazcko, C. Khanna

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A.K. LeBlanc, C.N. Mazcko

Study supervision: A.K. LeBlanc

The authors thank Drs. James Gulley, Michael Henson, Doug Thamm, and Tim Fan for their contributions to this manuscript.

This work was supported in part by the Intramural Research Program of the NIH, NCI.

1.
Paoloni
M
,
Khanna
C
. 
Translation of new cancer treatments from pet dogs to humans
.
Nat Rev Cancer
2008
;
8
:
147
56
.
2.
Rowell
JL
,
McCarthy
DO
,
Alvarez
CE
. 
Dog models of naturally occurring cancer
.
Trends Mol Med
2011
;
17
:
380
8
.
3.
Hansen
K
,
Khanna
C
. 
Spontaneous and genetically engineered animal models; use in preclinical cancer drug development
.
Eur J Cancer
2004
;
40
:
858
80
.
4.
Vail
DM
,
MacEwen
EG
. 
Spontaneously occurring tumors of companion animals as models for human cancer
.
Cancer Invest
2000
;
18
:
781
92
.
5.
Lairmore
MD
,
Khanna
C
. 
Naturally occurring diseases in animals: contributions to translational medicine
.
ILAR J
2014
;
55
:
1
3
.
6.
Gordon
I
,
Paoloni
M
,
Mazcko
C
,
Khanna
C
. 
The Comparative Oncology Trials Consortium: using spontaneously occurring cancers in dogs to inform the cancer drug development pathway
.
PLoS Med
2009
;
6
:
e1000161
.
7.
Paoloni
MC
,
Tandle
A
,
Mazcko
C
,
Hanna
E
,
Kachala
S
,
LeBlanc
A
, et al
Launching a novel preclinical infrastructure: comparative oncology trials consortium directed therapeutic targeting of TNFalpha to cancer vasculature
.
PLoS One
2009
;
4
:
e4972
.
8.
Khanna
C
,
London
C
,
Vail
D
,
Mazcko
C
,
Hirschfeld
S
. 
Guiding the optimal translation of new cancer treatments from canine to human cancer patients
.
Clin Cancer Res
2009
;
15
:
5671
7
.
9.
Gordon
I
,
Khanna
C
. 
Modeling opportunities in comparative oncology for drug development
.
ILAR J
2010
;
51
:
214
20
.
10.
London
CA
,
Bear
MD
,
McCleese
J
,
Foley
KP
,
Paalangara
R
,
Inoue
T
, et al
Phase I evaluation of STA-1474, a prodrug of the novel HSP90 inhibitor ganetespib, in dogs with spontaneous cancer
.
PLoS One
2011
;
6
:
e27018
.
11.
London
CA
,
Bernabe
LF
,
Barnard
S
,
Kisseberth
WC
,
Borgatti
A
,
Henson
M
, et al
Evaluation of the novel, orally bioavailable selective inhibitor of nuclear export (SINE) KPT-335 in spontaneous canine cancer: results of a Phase I study
.
PLoS One
2014
;
9
:
e87585
.
12.
Honigberg
LA
,
Smith
AM
,
Sirisawad
M
,
Verner
E
,
Loury
D
,
Chang
B
, et al
The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy
.
Proc Natl Acad Sci U S A
2010
;
107
:
13075
80
.
13.
Smith
MR
. 
Ibrutinib in B lymphoid malignancies
.
Expert Opin Pharmacother
2015
;
16
:
1879
87
.
14.
Barnard
RA
,
Wittenberg
LA
,
Amaravadi
RK
,
Gustafson
DL
,
Thorburn
A
,
Thamm
DH
. 
Phase I clinical trial and pharmacodynamic evaluation of combination hydroxychloroquine and doxorubicin treatment in pet dogs treated for spontaneously occurring lymphoma
.
Autophagy
2014
;
10
:
1
11
.
15.
.
Paoloni
M
,
Mazcko
C
,
Selting
K
,
Lana
S
,
Barber
L
,
Phillips
J
, et al
Defining the pharmacodynamic profile and therapeutic index of NHS-IL12 immunocytokine in dogs with malignant melanoma
.
PLoS One
2015
;
10
:
e0129954
.
16.
Paoloni
M
,
Davis
S
,
Lana
S
,
Withrow
S
,
Sangiorgi
L
,
Picci
P
, et al
Canine tumor cross-species genomics uncovers targets linked to osteosarcoma progression
.
BMC Genomics
2009
;
10
:
625
.
17.
Fidel
J
,
Kennedy
KC
,
Dernell
WS
,
Hansen
S
,
Wiss
V
,
Stroud
MR
, et al
Preclinical validation of the utility of BLZ-100 in providing fluorescence contrast for imaging spontaneous solid tumors
.
Cancer Res
2015
;
75
:
4283
91
.
18.
Paoloni
MC
,
Mazcko
C
,
Fox
E
,
Fan
T
,
Lana
S
,
Kisseberth
W
, et al
Rapamycin pharmacokinetic and pharmacodynamic relationships in osteosarcoma: a comparative oncology study in dogs
.
PLoS One
2010
;
5
:
e11013
.
19.
Richards
KL
,
Motsinger-Reif
AA
,
Chen
HW
,
Fedoriw
Y
,
Fan
C
,
Nielsen
DM
, et al
Gene profiling of canine B-cell lymphoma reveals germinal center and postgerminal center subtypes with different survival times, modeling human DLBCL
.
Cancer Res
2013
;
73
:
5029
39
.
20.
Mochizuki
H
,
Kennedy
K
,
Shapiro
SG
,
Breen
M
. 
BRAF mutations in canine cancers
.
PLoS One
2015
;
10
:
e0129534
.
21.
Roode
SC
,
Rotroff
D
,
Avery
AC
,
Suter
SE
,
Bienzle
D
,
Schiffman
JD
, et al
Genome-wide assessment of recurrent genomic imbalances in canine leukemia identifies evolutionarily conserved regions for subtype differentiation
.
Chromosome Res
. 
2015
.
[Epub ahead of print]
22.
Schiffman
JD
,
Breen
M
. 
Comparative oncology: what dogs and other species can teach us about humans with cancer
.
Philos Trans R Soc Lond B Biol Sci
2015
;
370
(1673).
23.
Pryer
NK
,
Lee
LB
,
Zadovaskaya
R
,
Yu
X
,
Sukbuntherng
J
,
Cherrington
JM
, et al
Proof of target for SU11654: inhibition of KIT phosphorylation in canine mast cell tumors
.
Clin Cancer Res
2003
;
9
:
5729
34
.
24.
London
CA
,
Malpas
PB
,
Wood-Follis
SL
,
Boucher
JF
,
Rusk
AW
,
Rosenberg
MP
, et al
Multi-center, placebo-controlled, double-blind, randomized study of oral toceranib phosphate (SU11654), a receptor tyrosine kinase inhibitor, for the treatment of dogs with recurrent (either local or distant) mast cell tumor following surgical excision
.
Clin Cancer Res
2009
;
15
:
3856
65
.
25.
Fowles
JS
,
Denton
CL
,
Gustafson
DL
Comparative analysis of MAPK and PI3K/AKT pathway activation and inhibition in human and canine melanoma
.
Vet Comp Oncol
2015
;
13
:
288
304
.
26.
Mazcko
CM
,
Thomas
R
. 
The establishment of the Pfizer-Canine Comparative Oncology and Genomics Consortium Biospecimen Repository
.
Vet Sci
2015
;
2
:
127
130
.
27.
Webster
JD
,
Simpson
ER
,
Michalowski
AM
,
Hoover
SB
,
Simpson
RM
. 
Quantifying histological features of cancer biospecimens for biobanking quality assurance using automated morphometric pattern recognition image analysis algorithms
J Biomol Tech
2011
;
22
:
108
18
.
28.
Liao
JC
,
Gregor
P
,
Wolchok
JD
,
Orlandi
F
,
Craft
D
,
Leung
C
, et al
Vaccination with human tyrosinase DNA induces antibody responses in dogs with advanced melanoma
.
Cancer Immun
2006
;
6
:
8
.
29.
Grosenbaugh
DA
,
Leard
AT
,
Bergman
PJ
,
Klein
MK
,
Meleo
K
,
Susaneck
S
, et al
Safety and efficacy of a xenogeneic DNA vaccine encoding for human tyrosinase as adjunctive treatment for oral malignant melanoma in dogs following surgical excision of the primary tumor
.
Am J Vet Res
2011
;
72
:
1631
8
.
30.
Andersen
BM
,
Pluhar
GE
,
Seiler
CE
,
Goulart
MR
,
SantaCruz
KS
,
Schutten
MM
, et al
Vaccination for invasive canine meningioma induces in situ production of antibodies capable of antibody-dependent cell-mediated cytoxicity
.
Cancer Res
2013
;
73
:
2987
97
.
31.
Turek
MM
,
Thamm
DH
,
Mitzey
A
,
Kurzman
ID
,
Huelsmeyer
MK
,
Dubielzig
RR
, et al
Human granulocyte-macrophage colony-stimulating factor DNA cationic-lipid complexed autologous tumour cell vaccination in the treatment of canine B-cell multicentric lymphoma
.
Vet Comp Oncol
2007
;
5
:
219
31
.
32.
U'Ren
LW
,
Biller
BJ
,
Elmslie
RE
,
Thamm
DH
,
Dow
SW
. 
Evaluation of a novel tumor vaccine in dogs with hemangiosarcoma
.
J Vet Intern Med
2007
;
21
:
113
20
.
33.
Laborda
E
,
Puig-Saus
C
,
Rodriguez-García
A
,
Moreno
R
,
Cascalló
M
,
Pastor
J
, et al
A pRb-responsive, RGD-modified, and hyaluronidase-armed canine oncolytic adenovirus for application in veterinary oncology
.
Mol Ther
2014
;
22
:
986
98
.
34.
Gentschev
I
,
Patil
SS
,
Petrov
I
,
Cappello
J
,
Adelfinger
M
,
Szalay
AA
. 
Oncolytic virotherapy of canine and feline cancer
.
Viruses
2014
;
6
:
2122
37
.
35.
Nguyen
SM
,
Thamm
DH
,
Vail
DM
,
London
CA
. 
Response evaluation criteria for solid tumours in dogs (v1.0): a Veterinary Cooperative Oncology Group (VCOG) consensus document
Vet Comp Oncol
2015
;
13
:
176
83
36.
Vail
DM
,
Michels
GM
,
Khanna
C
,
Selting
KA
,
London
CA
. 
Veterinary Cooperative Oncology Group Response evaluation criteria for peripheral nodal lymphoma in dogs (v1.0)–a Veterinary Cooperative Oncology Group (VCOG) consensus document
Vet Comp Oncol
2010
;
8
:
28
37
.
37.
Veterinary cooperative oncology group - common terminology criteria for adverse events (VCOG-CTCAE) following chemotherapy or biological antineoplastic therapy in dogs and cats v1.1
.
Vet Comp Oncol.
2011
Jul 20.
[Epub ahead of print]
.
38.
Thamm
DH
,
Vail
DM
,
Kurzman
ID
,
Babusis
D
,
Ray
AS
,
Sousa-Powers
N
, et al
GS-9219/VDC-1101–a prodrug of the acyclic nucleotide PMEG has antitumor activity in spontaneous canine multiple myeloma
.
BMC Vet Res
2014
;
10
:
30
.
39.
Vail
DM
,
Thamm
DH
,
Reiser
H
,
Ray
AS
,
Wolfgang
GH
,
Watkins
WJ
, et al
Assessment of GS-9219 in a pet dog model of non-Hodgkin's lymphoma
.
Clin Cancer Res
2009
;
15
:
3503
10
.
40.
LeBlanc
AK
,
Miller
AN
,
Galyon
GD
,
Moyers
TD
,
Long
MJ
,
Stuckey
AC
, et al
Preliminary evaluation of serial 18FDG-PET/CT to assess response to toceranib phosphate therapy in canine cancer
.
Vet Rad Ultrasound
2012
;
53
:
348
57
.
41.
Wittenburg
LA
,
Gustafson
DL
,
Thamm
DH
. 
Phase I pharmacokinetic and pharmacodynamic evaluation of combined valproic acid/doxorubicin treatment in dogs with spontaneous cancer.
Clin Cancer Res
2010
;
16
:
4832
42
.
42.
London
CA
,
Gardner
HL
,
Mathie
T
,
Stingle
N
,
Portela
R
,
Pennell
ML
, et al
Impact of toceranib/piroxicam/cyclophosphamide maintenance therapy on outcome of dogs with appendicular osteosarcoma following amputation and carboplatin chemotherapy: a multi-institutional study
.
PLoS One
2015
;
10
:
e0124889
.
43.
Thamm
DH
,
Kurzman
ID
,
Clark
MA
,
Ehrhart
EJ
,
Kraft
SL
,
Gustafson
DL
, et al
Preclinical investigation of PEGylated tumor necrosis factor alpha in dogs with spontaneous tumors: Phase I evaluation
.
Clin Cancer Res
2010
;
16
:
1498
508
.
44.
Selting
KA
,
Wang
X
,
Gustafson
DL
,
Henry
CJ
,
Villamil
JA
,
McCaw
DL
, et al
Evaluation of satraplatin in dogs with spontaneously occurring malignant tumors
.
J Vet Intern Med
2011
;
25
:
909
15
.
45.
Rutteman
GR
,
Erich
SA
,
Mol
JA
,
Spee
B
,
Grinwis
GC
,
Fleckenstein
L
, et al
Safety and efficacy field study of artesunate for dogs with non-resectable tumours
.
Anticancer Res
2013
;
33
:
1819
27
.
46.
Peterson
QP
,
Hsu
DC
,
Novotny
CJ
,
West
DC
,
Kim
D
,
Schmit
JM
, et al
Discovery and canine preclinical assessment of a nontoxic procaspase-3-activating compound
.
Cancer Res
2014
;
70
:
7232
41
.
47.
Sarver
AL
,
Thayanithy
V
,
Scott
MC
,
Cleton-Jansen
AM
,
Hogendoorn
PC
,
Modiano
JF
, et al
MicroRNAs at the human 14q32 locus have prognostic significance in osteosarcoma
.
Orphanet J Rare Dis
2013
;
8
:
7
.
48.
Stratham-Ringen
KA
,
Selting
KA
,
Lattimer
JC
,
Henry
CJ
,
Green
JA
,
Bryan
JN
, et al
Evaluation of a B-cell leukemia-lymphoma 2-specific radiolabeled peptide nucleic acid-peptide conjugate for scintigraphic detection of neoplastic lymphocytes in dogs with B-cell lymphoma
.
Am J Vet Res
2012
;
73
:
681
688
.
49.
Zwingenberger
AL
,
Kent
MS
,
Liu
R
,
Kukis
DL
,
Wisner
ER
,
DeNardo
SJ
, et al
In-vivo biodistribution and safety of 99mTc-LLP2A-HYNIC in canine non-Hodgkin lymphoma
.
PLoS One
2012
;
7
:
e34404
.
50.
Bradshaw
TJ
,
Bowen
SR
,
Jallow
N
,
Forrest
LJ
,
Jeraj
R
. 
Heterogeneity in intratumor correlations of 18F-FDG, 18F-FLT, and 61Cu-ATSM PET in canine sinonasal tumors
.
J Nucl Med
2013
;
54
:
1931
7
.
51.
Hansen
AE
,
Kristensen
AT
,
Law
I
,
McEvoy
FJ
,
Kjaer
A
,
Engelholm
SA
. 
Multimodality functional imaging of spontaneous canine tumors using 64Cu-ATSM and 18FDG PET/CT and dynamic contrast enhanced perfusion CT
.
Radiother Oncol
2012
;
102
:
424
8
.
52.
Hansen
AE
,
Kristensen
AT
,
Jorgensen
JT
,
McEvoy
FJ
,
Busk
M
,
van der Kogel
AJ
, et al
64Cu-ATSM and 18FDG PET uptake and 64Cu-ATSM autoradiography in spontaneous canine tumors: comparison with pimonidazole hypoxia immunohistochemistry
.
Radiat Oncol
2012
;
7
:
89
.
53.
Black
NF
,
McJames
S
,
Kadrmas
DJ
. 
Rapid multi-tracer PET tumor imaging with 18F-FDG and secondary short-lived tracers
.
IEEE Trans Nucl Sci
2009
;
56
:
2750
8
.
54.
Black
NF
,
McJames
S
,
Rust
TC
,
Kadrmas
DJ
. 
Evaluation of rapid dual-tracer 62Cu-PTSM + 62Cu-ATSM PET in dogs with spontaneously-occurring tumors
.
Phys Med Biol
2008
;
53
:
217
32
.
55.
Khanna
C
,
Fan
TM
,
Gorlick
R
,
Helman
LJ
,
Kleinerman
ES
,
Adamson
PC
, et al
Toward a drug development path that targets metastatic progression in osteosarcoma
.
Clin Cancer Res
2014
;
20
:
4200
9
.
56.
Paoloni
M
,
Webb
C
,
Mazcko
C
,
Cherba
D
,
Hendricks
W
,
Lana
S
, et al
Prospective molecular profiling of canine cancers provides a clinically relevant comparative model for evaluating personalized medicine (PMed) trials
.
PLoS One
2014
;
9
:
e90028
.
57.
Modiano
JF
,
Bellgrau
D
,
Cutter
GR
,
Lana
SE
,
Ehrhart
NP
,
Ehrhart
E
, et al
Inflammation, apoptosis, and necrosis induced by neoadjuvant Fas ligand gene therapy improves survival of dogs with spontaneous bone cancer
.
Mol Ther
2012
;
20
:
2234
43
.